Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Gene Ther ; 30(5): 455-462, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-33608675

RESUMO

Clostridium difficile is the leading cause of antibiotic-associated nosocomial diarrhea in the developed world. When the host-associated colon microbiome is disrupted by the ingestion of antibiotics, C. difficile spores can germinate, resulting in infection. C. difficile secretes enterotoxin A (TcdA) and cytotoxin B (TcdB) that are responsible for disease pathology. Treatment options are limited as the bacterium demonstrates resistance to many antibiotics, and even with antibacterial therapies, recurrences of C. difficile are common. Actotoxumab and bezlotoxumab are human monoclonal antibodies that bind and neutralize TcdA and TcdB, respectively. In 2016, the US food and drug administration (FDA) approved bezlotoxumab for use in the prevention of C. difficile infection recurrence. To ensure the long-term expression of antibodies, gene therapy can be used. Here, adeno-associated virus (AAV)6.2FF, a novel triple mutant of AAV6, was engineered to express either actotoxumab or bezlotoxumab in mice and hamsters. Both antibodies expressed at greater than 90 µg/mL in the serum and were detected at mucosal surfaces in both models. Hundred percent of mice given AAV6.2FF-actoxumab survived a lethal dose of TcdA. This proof of concept study demonstrates that AAV-mediated expression of C. difficile toxin antibodies is a viable approach for the prevention of recurrent C. difficile infections.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Infecções por Clostridium , Humanos , Animais , Camundongos , Toxinas Bacterianas/genética , Anticorpos Neutralizantes , Infecções por Clostridium/prevenção & controle , Infecções por Clostridium/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Antibacterianos/uso terapêutico , Proteínas de Bactérias/genética , Proteínas de Bactérias/uso terapêutico
2.
Int J Mol Sci ; 23(9)2022 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-35563458

RESUMO

Cutibacterium acnes (C. acnes) has been implicated in inflammatory acne where highly mutated Christie-Atkins-Munch-Petersen factor (CAMP)1 displays strong toll like receptor (TLR)-2 binding activity. Using specific antibodies, we showed that CAMP1 production was independent of C. acnes phylotype and involved in the induction of inflammation. We confirmed that TLR-2 bound both mutated and non-mutated recombinant CAMP1, and peptide array analysis showed that seven peptides (A14, A15, B1, B2, B3, C1 and C3) were involved in TLR-2 binding, located on the same side of the three-dimensional structure of CAMP1. Both mutated and non-mutated recombinant CAMP1 proteins induced the production of C-X-C motif chemokine ligand interleukin (CXCL)8/(IL)-8 in vitro in keratinocytes and that of granulocyte macrophage-colony stimulating factor (GM-CSF), tumor necrosis factor (TNF)-α, IL-1ß and IL-10 in ex vivo human skin explants. Only A14, B1 and B2 inhibited the production of CXCL8/IL-8 by keratinocytes and that of (GM-CSF), TNF-α, IL-1ß and IL-10 in human skin explants stimulated with rCAMP1 and C. acnes. Following pretreatment with B2, RNA sequencing on skin explants identified the 10 genes displaying the strongest differential expression as IL6, TNF, CXCL1, CXCL2, CXCL3, CXCL8, IL-1ß, chemokine ligand (CCL)2, CCL4 and colony stimulating factor (CSF)2. We, thus, identified a new CAMP1-derived peptide as a TLR-2 modulator likely to be a good candidate for clinical evaluation.


Assuntos
Proteínas de Bactérias , Inflamação , Propionibacteriaceae , Receptor 2 Toll-Like , Proteínas de Bactérias/farmacologia , Proteínas de Bactérias/uso terapêutico , Quimiocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-10/metabolismo , Ligantes , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Propionibacteriaceae/química , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Biochimie ; 192: 83-90, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34653542

RESUMO

Bacillus thuringiensis (Bt) is a ubiquitous bacterium that produces several proteins that are toxic to different invertebrates such as insects, nematodes, mites, and also some protozoans. Among these, Cry and Cyt proteins are most explored as biopesticides for their action against agricultural pests and vectors of human diseases. In 2000, a group of researchers from Japan isolated parasporal inclusion proteins from B. thuringiensis, and reported their cytotoxic action against human leukemia. Later, other proteins with similar antitumor properties were also isolated from this bacterium and these cytotoxic proteins with specific activity against human cancer cells were named parasporins. At present, nineteen different parasporins are registered and classified in six families. These parasporins have been described to have specific in vitro antitumor activity against several cancer cell lines. The antitumor activity makes parasporins possible candidates as anticancer agents. Various research groups around the world are involved in isolating and characterizing in vitro antitumor activity of these proteins and many articles reporting such activities in detail have been published. However, there are virtually no data regarding the antitumor activity of parasporins in vivo. This review summarizes the properties of these potentially useful antitumor agents of natural origin, focusing on their in vivo activity thus also highlighting the importance of testing these proteins in animal models for a possible application in clinical oncology.


Assuntos
Bacillus thuringiensis/química , Proteínas de Bactérias , Citotoxinas , Endotoxinas , Corpos de Inclusão Intranuclear/química , Leucemia/tratamento farmacológico , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/uso terapêutico , Linhagem Celular Tumoral , Citotoxinas/química , Citotoxinas/uso terapêutico , Endotoxinas/química , Endotoxinas/uso terapêutico , Humanos , Leucemia/metabolismo , Controle Biológico de Vetores
4.
Int Urol Nephrol ; 54(7): 1681-1691, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34783980

RESUMO

PURPOSE: To evaluate the effects of manganese superoxide dismutase (Mn-SOD) from thermophilic bacterium HB27 (name as Tt-SOD) on chemical cystitis. METHODS: Control and experimental rats were infused by intravesical saline or hydrochloric acid (HCl) on the first day of the experiments. Saline, sodium hyaluronate (SH) or Tt-SOD were infused intravesically once a day for three consequent days. On the fifth day, the rats were weighted and sacrificed following a pain threshold test. The bladder was harvested for histological and biochemical analyses. RESULTS: Tt-SOD could reduce the bladder index, infiltration of inflammatory cells in tissues, serum inflammatory factors and SOD levels, mRNA expression of inflammatory factors in tissues, and increase perineal mechanical pain threshold and serum MDA and ROS levels in HCl-induced chemical cystitis. Furthermore, Tt-SOD alleviated inflammation and oxidative stress by the negative regulation of the NF-κB p65 and p38 MAPK signaling pathway. CONCLUSIONS: Intravesical instillation of Tt-SOD provides protective effects against HCl-induced cystitis.


Assuntos
Proteínas de Bactérias , Cistite , Superóxido Dismutase , Animais , Proteínas de Bactérias/uso terapêutico , Cistite/induzido quimicamente , Cistite/terapia , Ácido Clorídrico/efeitos adversos , Inflamação/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/uso terapêutico , Bexiga Urinária/patologia
5.
J Clin Invest ; 131(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34196308

RESUMO

Clinical immunotherapy approaches are lacking efficacy in the treatment of glioblastoma (GBM). In this study, we sought to reverse local and systemic GBM-induced immunosuppression using the Helicobacter pylori neutrophil-activating protein (NAP), a potent TLR2 agonist, as an immunostimulatory transgene expressed in an oncolytic measles virus (MV) platform, retargeted to allow viral entry through the urokinase-type plasminogen activator receptor (uPAR). While single-agent murine anti-PD1 treatment or repeat in situ immunization with MV-s-NAP-uPA provided modest survival benefit in MV-resistant syngeneic GBM models, the combination treatment led to synergy with a cure rate of 80% in mice bearing intracranial GL261 tumors and 72% in mice with CT-2A tumors. Combination NAP-immunovirotherapy induced massive influx of lymphoid cells in mouse brain, with CD8+ T cell predominance; therapeutic efficacy was CD8+ T cell dependent. Inhibition of the IFN response pathway using the JAK1/JAK2 inhibitor ruxolitinib decreased PD-L1 expression on myeloid-derived suppressor cells in the brain and further potentiated the therapeutic effect of MV-s-NAP-uPA and anti-PD1. Our findings support the notion that MV strains armed with bacterial immunostimulatory antigens represent an effective strategy to overcome the limited efficacy of immune checkpoint inhibitor-based therapies in GBM, creating a promising translational strategy for this lethal brain tumor.


Assuntos
Antígenos de Bactérias/uso terapêutico , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/uso terapêutico , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/metabolismo , Efeito Citopatogênico Viral , Feminino , Glioblastoma/imunologia , Glioblastoma/patologia , Humanos , Linfócitos do Interstício Tumoral/imunologia , Vírus do Sarampo/genética , Vírus do Sarampo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Pesquisa Translacional Biomédica , Internalização do Vírus
6.
J Gastrointest Cancer ; 52(2): 593-599, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32524303

RESUMO

INTRODUCTION: Cancer is the uncontrolled division of cells and can be caused by genetic or environmental factors. Pancreatic cancer is one of the deadliest among all cancers. The role of bacteria as an anticancer agent dates back to almost 100 years ago. The microbiome has recently become a focus of research in carcinogenesis and even pancreatic cancer. Shigella flexneri is a gram-negative bacterium, which causes shigellosis with symptoms such as diarrhea, fever, and stomach cramps in human. Shigella flexneri may play a very important role in the internal pathways of apoptosis and may induce apoptosis in some of the cancerous cells. MATERIAL AND METHODS: In this experiment bacteria were cultured on Salmonella-Shigella agar, then inoculated into BHI Broth medium. After sonication, the protein concentration of the bacterium was measured by using the ZellBio Sensitive Protein Bradford Assay kit. MTT assay was performed to obtain IC50 for the said bacterial protein. Later by cDNA kit synthesized the cDNA based on the RNA template. In the end, the results were analyzed using real-time PCR and the expression of Bax and Bcl-2 genes was measured before and after treatment. RESULTS: The results showed that Shigella flexneri has the potential anti-proliferative effect in pancreatic cancer. The inhibitory concentration, pro-apoptotic amount to upregulate Bax, and meanwhile also to downregulate the bcl-2 found to be 10 µl. CONCLUSION: In general, due to defects in the apoptotic pathway in cancer cells and the existence of drug-resistant cells, the detection of new apoptotic inducers such as Shigella flexneri cell extract can be used for further studies on cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Fatores Biológicos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Shigella flexneri , Apoptose/genética , Proteínas de Bactérias/uso terapêutico , Fatores Biológicos/uso terapêutico , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Regulação para Cima/efeitos dos fármacos , Proteína X Associada a bcl-2/genética
7.
Transplantation ; 105(8): 1808-1817, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33093408

RESUMO

BACKGROUND: Highly HLA sensitized patients have limited access to life-saving kidney transplantation because of a paucity of immunologically suitable donors. Imlifidase is a cysteine protease that cleaves IgG leading to a rapid decrease in antibody level and inhibition of IgG-mediated injury. This study investigates the efficacy and safety of imlifidase in converting a positive crossmatch test to negative, allowing highly sensitized patients to be transplanted with a living or deceased donor kidney. METHODS: This open-label, single-arm, phase 2 trial conducted at 5 transplant centers, evaluated the ability of imlifidase to create a negative crossmatch test within 24 h. Secondary endpoints included postimlifidase donor-specific antibody levels compared with predose levels, renal function, and pharmacokinetic/pharmacodynamic profiles. Safety endpoints included adverse events and immunogenicity profile. RESULTS: Of the transplanted patients, 89.5% demonstrated conversion of baseline positive crossmatch to negative within 24 h after imlifidase treatment. Donor-specific antibodies most often rebounded 3-14 d postimlifidase dose, with substantial interpatient variability. Patient survival was 100% with graft survival of 88.9% at 6 mo. With this, 38.9% had early biopsy proven antibody-mediated rejection with onset 2-19 d posttransplantation. Serum IgG levels began to normalize after ~3-7 d posttransplantation. Antidrug antibody levels were consistent with previous studies. Seven adverse events in 6 patients were classified as possibly or probably related to treatment and were mild-moderate in severity. CONCLUSIONS: Imlifidase was well tolerated, converted positive crossmatches to negative, and enabled patients with a median calculated panel-reactive antibody of 99.83% to undergo kidney transplantation resulting in good kidney function and graft survival at 6 mo.


Assuntos
Proteínas de Bactérias/uso terapêutico , Dessensibilização Imunológica/métodos , Teste de Histocompatibilidade , Transplante de Rim , Adulto , Proteínas de Bactérias/efeitos adversos , Feminino , Rejeição de Enxerto , Sobrevivência de Enxerto , Humanos , Isoanticorpos/sangue , Masculino , Pessoa de Meia-Idade , Doadores de Tecidos , Adulto Jovem
8.
Drugs ; 80(17): 1859-1864, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33058042

RESUMO

Imlifidase (IdefirixTM), a cysteine protease derived from the immunoglobulin G (IgG)­degrading enzyme of Streptococcus (S.) pyogenes is being developed by Hansa Biopharma AB for treatment of transplant rejection and rare IgG-mediated autoimmune conditions. In August 2020, intravenous imlifidase received its first global approval in the EU for desensitization treatment of highly sensitized adult kidney transplant patients with positive crossmatch against an available deceased donor. Imlifidase is currently undergoing clinical evaluation for the prevention of kidney transplant rejection in the USA, Australia, France and Austria, and clinical development is underway for anti-glomerular basement membrane disease, and for Guillain-Barre syndrome in France, the UK and the Netherlands. This article summarizes the milestones in the development of imlifidase leading to this first approval for desensitization treatment of highly sensitized adult kidney transplant patients with positive crossmatch against an available deceased donor.


Assuntos
Doença Antimembrana Basal Glomerular/tratamento farmacológico , Proteínas de Bactérias/uso terapêutico , Aprovação de Drogas , Rejeição de Enxerto/tratamento farmacológico , Síndrome de Guillain-Barré/tratamento farmacológico , Administração Intravenosa , Animais , Doença Antimembrana Basal Glomerular/imunologia , Proteínas de Bactérias/farmacologia , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Dessensibilização Imunológica/métodos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , União Europeia , Síndrome de Guillain-Barré/imunologia , Antígenos HLA/imunologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Falência Renal Crônica/cirurgia , Transplante de Rim/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Doenças Raras/tratamento farmacológico , Doenças Raras/imunologia
9.
Biochimie ; 177: 142-152, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32828824

RESUMO

Phenylalanine ammonia lyase (PAL) has recently emerged as an important therapeutic enzyme with several biomedical applications. The enzyme catabolizes l-phenylalanine to trans-cinnamate and ammonia. PAL is widely distributed in higher plants, some algae, ferns, and microorganisms, but absent in animals. Although microbial PAL has been extensively exploited in the past for producing industrially important metabolites, its high substrate specificity and catalytic efficacy lately spurred interest in its biomedical applications. PEG-PAL drug named Palynziq™, isolated from Anabaena variabilis has been recently approved for the treatment of adult phenylketonuria (PKU) patients. Further, it has exhibited high potency in regressing tumors and treating tyrosine related metabolic abnormalities like tyrosinemia. Several therapeutically valuable metabolites have been biosynthesized via its catalytic action including dietary supplements, antimicrobial peptides, aspartame, amino-acids, and their derivatives. This review focuses on all the prospective biomedical applications of PAL. It also provides an overview of the structure, production parameters, and various strategies to improve the therapeutic potential of this enzyme. Engineered PAL with improved pharmacodynamic and pharmacokinetic properties will further establish this enzyme as a highly efficient biological drug.


Assuntos
Proteínas de Bactérias/farmacologia , Proteínas de Bactérias/uso terapêutico , Fenilalanina Amônia-Liase/farmacologia , Fenilalanina Amônia-Liase/uso terapêutico , Erros Inatos do Metabolismo dos Aminoácidos/tratamento farmacológico , Animais , Anti-Infecciosos/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Suplementos Nutricionais , Humanos , Neoplasias/tratamento farmacológico , Fenilalanina Amônia-Liase/química , Fenilalanina Amônia-Liase/genética
10.
In Vivo ; 34(3 Suppl): 1593-1596, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32503816

RESUMO

The Covid-19 pandemic is a world-wide crisis without an effective therapy. While most approaches to therapy are using repurposed drugs that were developed for other diseases, it is thought that targeting the biology of the SARS-CoV-2 virus, which causes Covid-19, can result in an effective therapeutic treatment. The coronavirus RNA cap structure is methylated by two viral methyltransferases that transfer methyl groups from S-adenosylmethionine (SAM). The proper methylation of the virus depends on the level of methionine in the host to form SAM. Herein, we propose to restrict methionine availability by treating the patient with oral recombinant methioninase, aiming to treat Covid-19. By restricting methionine we not only interdict viral replication, which depends on the viral RNA cap methyaltion, but also inhibit the proliferation of the infected cells, which have an increased requirement for methionine. Most importantly, the virally-induced T-cell- and macrophage-mediated cytokine storm, which seems to be a significant cause for Covid-19 deaths, can also be inhibited by restricting methionine, since T-cell and macrophrage activation greatly increases the methionine requirement for these cells. The evidence reviewed here suggests that oral recombinant methioninase could be a promising treatment for coronavirus patients.


Assuntos
Antivirais/uso terapêutico , Betacoronavirus/efeitos dos fármacos , Liases de Carbono-Enxofre/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Metionina/metabolismo , Pneumonia Viral/tratamento farmacológico , Capuzes de RNA/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Administração Oral , Antivirais/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/uso terapêutico , Betacoronavirus/fisiologia , COVID-19 , Liases de Carbono-Enxofre/administração & dosagem , Ensaios Clínicos como Assunto , Infecções por Coronavirus/complicações , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/prevenção & controle , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Metanálise como Assunto , Metilação/efeitos dos fármacos , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/imunologia , Pseudomonas putida/enzimologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/uso terapêutico , S-Adenosilmetionina/metabolismo , SARS-CoV-2 , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Replicação Viral/efeitos dos fármacos
11.
Recent Pat Biotechnol ; 14(3): 235-246, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32208128

RESUMO

BACKGROUND: Arginine deiminase is a bacterial enzyme, which degrades L-arginine. Some human cancers such as hepatocellular carcinoma (HCC) and melanoma are auxotrophic for arginine. Therefore, PEGylated arginine deiminase (ADI-PEG20) is a good anticancer candidate with antitumor effects. It causes local depletion of L-arginine and growth inhibition in arginineauxotrophic tumor cells. The FDA and EMA have granted orphan status to this drug. Some recently published patents have dealt with this enzyme or its PEGylated form. OBJECTIVE: Due to increasing attention to it, we aimed to evaluate and compare 30 arginine deiminase proteins from different bacterial species through in silico analysis. METHODS: The exploited analyses included the investigation of physicochemical properties, multiple sequence alignment (MSA), motif, superfamily, phylogenetic and 3D comparative analyses of arginine deiminase proteins thorough various bioinformatics tools. RESULTS: The most abundant amino acid in the arginine deiminase proteins is leucine (10.13%) while the least amino acid ratio is cysteine (0.98%). Multiple sequence alignment showed 47 conserved patterns between 30 arginine deiminase amino acid sequences. The results of sequence homology among 30 different groups of arginine deiminase enzymes revealed that all the studied sequences located in amidinotransferase superfamily. Based on the phylogenetic analysis, two major clusters were identified. Considering the results of various in silico studies; we selected the five best candidates for further investigations. The 3D structures of the best five arginine deiminase proteins were generated by the I-TASSER server and PyMOL. The RAMPAGE analysis revealed that 81.4%-91.4%, of the selected sequences, were located in the favored region of arginine deiminase proteins. CONCLUSION: The results of this study shed light on the basic physicochemical properties of thirty major arginine deiminase sequences. The obtained data could be employed for further in vivo and clinical studies and also for developing the related therapeutic enzymes.


Assuntos
Antineoplásicos/química , Arginina/metabolismo , Bactérias/enzimologia , Proteínas de Bactérias/química , Hidrolases/química , Polietilenoglicóis/química , Sequência de Aminoácidos , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Arginina/química , Bactérias/classificação , Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Biologia Computacional/métodos , Simulação por Computador , Sequência Conservada , Expressão Gênica , Humanos , Hidrolases/genética , Hidrolases/metabolismo , Hidrolases/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Modelos Moleculares , Patentes como Assunto , Filogenia , Polietilenoglicóis/metabolismo , Polietilenoglicóis/uso terapêutico , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo
12.
Biologicals ; 64: 28-33, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31959505

RESUMO

Erwinia chrysanthemil-asparaginase (ErA) has been used for the treatment of acute lymphoblastic leukaemia (ALL) for decades, and its safety and efficacy have been well demonstrated. ErA drug substance and drug product contain a small proportion of acidic isoforms, with a known mechanism of formation, which have been shown to be minor conformational variants retaining enzymatic activity and function. Specifications for these acidic isoforms were set with an extremely limited data set, and with further manufacturing experience, it can now be demonstrated that they were set too tightly. Here, we consider the ability of the manufacturing process to meet the current acidic isoforms specifications, as well as clinical outcomes from drug product containing a higher proportion of isoforms. Compared with the historical clinical experience with the drug, there appeared to be no difference in the rate of adverse event reporting (e.g., hypersensitivity or other events) when drug product with relatively higher acidic isoforms was administered. ErA acidic isoforms comprise part of the ErA product and appear to have no clinical relevance, so a realignment of process capability and specification may be warranted. Biopharmaceutical developers should exercise caution when setting specifications with limited data, to avoid process capability pitfalls later.


Assuntos
Asparaginase/química , Proteínas de Bactérias/química , Dickeya chrysanthemi/enzimologia , Asparaginase/uso terapêutico , Proteínas de Bactérias/uso terapêutico , Humanos , Isoenzimas/química , Isoenzimas/uso terapêutico
13.
Int J Biol Macromol ; 136: 27-34, 2019 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-31185242

RESUMO

Lactobacillus acidophilus NCFM, a probiotic generally regarded as safe, carries a proteinaceous surface (S) layer, composed of numerous identical subunits (surface layer protein, Slp). S-layer proteins have been confirmed to possess multiple biological properties, but their role in maintaining the intestinal epithelial barrier is not fully known. We investigated the effects of Slp on tumor necrosis factor (TNF)-α-elicited intestinal barrier dysfunction and explored the underlying molecular mechanism. TNF-α administration markedly induced intestinal epithelial injury and inflammation in Caco-2 cells. Preincubation of Caco-2 cells with Slp at concentrations ranging from 50 to 100 µg/mL for 6 h improved intestinal epithelial cell integrity and permeability, restored ZO-1 and Occludin protein expressions (P < 0.05) and reduced the secretion of interleukin 8 by a maximum of 47.8%. Furthermore, the addition of Slp to Caco-2 cell monolayers attenuated cell apoptosis and inhibited nuclear factor-κB (NF-κB) p65 nucleus translocation by suppressing the activation of NF-κB. Collectively, the ability of Slp to attenuate dysfunction of the intestinal epithelial barrier stimulated by TNF-α and to exert anti-inflammatory effects supports its potential use in the development of functional foods and in the prevention of inflammatory bowel diseases.


Assuntos
Proteínas de Bactérias/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Lactobacillus acidophilus , Glicoproteínas de Membrana/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Proteínas de Bactérias/uso terapêutico , Células CACO-2 , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/tratamento farmacológico , Interleucina-8/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Glicoproteínas de Membrana/uso terapêutico , Permeabilidade/efeitos dos fármacos , Proteínas de Junções Íntimas/metabolismo , Fator de Transcrição RelA/metabolismo
14.
Cell Death Dis ; 10(4): 302, 2019 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-30944311

RESUMO

Severe side effects often restrict clinical application of the widely used chemotherapeutic drug doxorubicin. In order to decrease required substance concentrations, new concepts for successful combination therapy are needed. Since doxorubicin causes DNA damage, combination with compounds that modulate DNA repair could be a promising strategy. Very recently, a role of nuclear actin for DNA damage repair has been proposed, making actin a potential target for cancer therapy in combination with DNA-damaging therapeutics. This is of special interest, since actin-binding compounds have not yet found their way into clinics. We find that low-dose combination treatment of doxorubicin with the actin polymerizer chondramide B (ChB) synergistically inhibits tumor growth in vivo. On the cellular level we demonstrate that actin binders inhibit distinctive double strand break (DSB) repair pathways. Actin manipulation impairs the recruitment of replication factor A (RPA) to the site of damage, a process crucial for homologous recombination. In addition, actin binders reduce autophosphorylation of DNA-dependent protein kinase (DNA-PK) during nonhomologous end joining. Our findings substantiate a direct involvement of actin in nuclear DSB repair pathways, and propose actin as a therapeutic target for combination therapy with DNA-damaging agents such as doxorubicin.


Assuntos
Actinas/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteínas de Bactérias/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Depsipeptídeos/uso terapêutico , Doxorrubicina/uso terapêutico , Actinas/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proteínas de Bactérias/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Morte Celular/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Proteína Quinase Ativada por DNA/metabolismo , Depsipeptídeos/farmacologia , Doxorrubicina/farmacologia , Células HeLa , Humanos , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Fosforilação , Recombinação Genética/efeitos dos fármacos , Proteína de Replicação A/genética , Proteína de Replicação A/metabolismo , Tiazolidinas/farmacologia , Tiazolidinas/uso terapêutico , Transplante Heterólogo
15.
Biologicals ; 59: 47-55, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30871932

RESUMO

Acute lymphoblastic leukemia (ALL) is a type of cancer with a high incidence in children. The enzyme l-asparaginase (ASNase) constitutes a key element in the treatment of this disease. Four formulations of ASNase from a bacterial source are currently available. However, these formulations are characterized by their high immunogenicity, resulting in the inactivation of the drug, as well as in the occurrence of hypersensitivity reactions in a large number of patients. In this work, we performed an immunoinformatic analysis in order to clarify structural aspects of the immunogenicity of the asparaginase from Escherichia coli and Erwinia carotovora. For this purpose, we performed the prediction of immunogenic and allergenic epitopes in the structure of asparaginases by using the relative frequency of immunogenic peptides for the eight alleles most frequently distributed worldwide. This study showed that there are no significant differences in the level of immunogenicity between the two enzymes, while asparaginase from E. coli presented a higher relative frequency of allergenic epitopes. These results are consistent with previously published reports. However, from a structural point of view, to the best of our knowledge, this is the first report describing the structural determinants that contribute to the hypersensitivity response to this treatment.


Assuntos
Asparaginase/imunologia , Proteínas de Bactérias/imunologia , Epitopos/imunologia , Escherichia coli/enzimologia , Pectobacterium carotovorum/enzimologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Sequência de Aminoácidos , Asparaginase/efeitos adversos , Asparaginase/uso terapêutico , Proteínas de Bactérias/química , Proteínas de Bactérias/uso terapêutico , Criança , Simulação por Computador , Epitopos/química , Escherichia coli/genética , Humanos , Hipersensibilidade/etiologia , Hipersensibilidade/imunologia , Pectobacterium carotovorum/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Conformação Proteica , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
16.
Appl Microbiol Biotechnol ; 103(5): 2053-2066, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30656391

RESUMO

Lactic acid bacteria (LAB) have a long-term history of use in food industry and are becoming attractive for use in therapy on account of their safety, intrinsic beneficial health effects, and considerable biotechnological potential. The established systems for engineering are combined with novel approaches, such as CRISPR-Cas, to enable the use of LAB as vectors for delivery of various therapeutic molecules. The latter are either secreted or surface displayed and can be used for the treatment or prevention of numerous conditions: inflammatory bowel diseases, infections, autoimmune diseases, and even cancer. This review presents some recent data on engineering of LAB, with the emphasis on the most commonly used genera Lactococcus and Lactobacillus. Their use for the delivery of therapeutic proteins is discussed, while a special focus is given to the delivery of therapeutic peptides. Therapeutically relevant improvements of engineered LAB, such as containment systems, ability to visualize bacteria, or target specific host cells are also addressed. Future engineering of LAB for therapy will adopt the capabilities of synthetic biology, with first examples already emerging.


Assuntos
Produtos Biológicos/metabolismo , Produtos Biológicos/uso terapêutico , Lactobacillus/metabolismo , Lactococcus lactis/metabolismo , Engenharia de Proteínas/métodos , Proteínas de Bactérias/genética , Proteínas de Bactérias/uso terapêutico , Edição de Genes/métodos , Humanos , Lactobacillus/genética , Lactococcus lactis/genética
17.
Clin Transl Gastroenterol ; 9(9): 181, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30228265

RESUMO

INTRODUCTION: Recently, the population of individuals with non-celiac gluten sensitivity (NCGS) who do not have celiac disease but show improved symptoms with a gluten-free diet, has increased. Enzyme replacement therapy using digestive enzymes is expected to improve the symptoms of NCGS and be sustainable, since gluten-related proteins that are indigestible by the digestive system have been considered triggers of NCGS. METHODS: We selected patients with NCGS by screening demographic interviews, as well as performing medical evaluations, anti-gluten antibody tests, and gluten challenge tests. We performed a single-blind and crossover clinical trial with these subjects using a gluten challenge with the enzyme mixture or a placebo. Our designed enzyme mixture contained peptidase, semi alkaline protease, deuterolysin, and cysteine protease derived from Aspergillus oryzae, Aspergillus melleus, Penicillium citrinum, and Carica papaya L., respectively. RESULTS: Administration of the enzyme mixture significantly decreased the change in the score of the symptom questionnaire before and after the gluten challenge compared with administration of the placebo in patients with NCGS without adverse events. In particular, the changes in the score of the gluten-induced incomplete evacuation feeling and headaches were significantly improved. The serum levels of interleukin (IL)-8, tumor necrosis factor (TNF)-α, andregulated on activation, normal T cell expressed and secreted (RANTES) in subjects were not significantly changed by gluten, as expected from previous studies, and the enzyme mixture did not affect these inflammatory markers. CONCLUSION: In this human clinical study, we demonstrated the efficacy of the enzyme mixture derived from microorganisms and papaya in improving the symptoms of NCGS.


Assuntos
Terapia de Reposição de Enzimas , Hipersensibilidade Alimentar/diagnóstico , Hipersensibilidade Alimentar/terapia , Glutens/efeitos adversos , Adulto , Idoso , Aspergillus/enzimologia , Proteínas de Bactérias/uso terapêutico , Carica/enzimologia , Estudos Cross-Over , Cisteína Proteases/uso terapêutico , Citocinas/sangue , Dieta Livre de Glúten , Digestão , Endopeptidases/uso terapêutico , Feminino , Hipersensibilidade Alimentar/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Penicillium/enzimologia , Peptídeo Hidrolases/uso terapêutico , Método Simples-Cego , Adulto Jovem
18.
Biomed Pharmacother ; 101: 929-937, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29635902

RESUMO

Natural products may represent a rich source of new drugs. The enthusiasm toward this topic has recently been fueled by the 2015 Nobel Prize in Physiology or Medicine, awarded for the discovery of avermectin and artemisinin, natural products from Bacteria and Plantae, respectively, which have targeted one of the major global health issues, the parasitic diseases. Specifically, bacteria either living in the environment or colonizing our body may produce compounds of unexpected biomedical value with the potentiality to be employed as therapeutic drugs. In this review, the fascinating history of CNF1, a protein toxin produced by pathogenic strains of Escherichia coli, is divulged. Even if produced by bacteria responsible for a variety of diseases, CNF1 can behave as a promising benefactor to mankind. By modulating the Rho GTPases, this bacterial product plays a key role in organizing the actin cytoskeleton, enhancing synaptic plasticity and brain energy level, rescuing cognitive deficits, reducing glioma growth in experimental animals. These abilities strongly suggest the need to proceed with the studies on this odd drug in order to pave the way toward clinical trials.


Assuntos
Bactérias/química , Toxinas Bacterianas/uso terapêutico , Proteínas de Escherichia coli/uso terapêutico , Animais , Proteínas de Bactérias/uso terapêutico , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Humanos
19.
Mol Immunol ; 94: 176-182, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29324238

RESUMO

Nearly half of the world's population is infected with Helicobacter pylori. Clinical manifestations of this infection range from gastritis and peptic ulcers to gastric adenocarcinoma and lymphoma. Due to the emerging of antibiotic resistant strains and poor patient compliance of the antibiotic therapy, there is increasing interest in the development of a protective vaccine against H. pylori infection. The bacterial protein FliD forms a capping structure on the end of each flagellum which is critical to prevent depolymerization and structural degradation. In this study, the potential of FliD as a prospective H. pylori subunit vaccine was assessed. For this purpose, immunogenicity and protective efficacy of recombinant FliD (rFliD) from H. pylori was evaluated in C57BL/6 mice. Purified rFliD was formulated with different adjuvants and administered via subcutaneous or oral route. Subcutaneous immunization with rFliD elicited predominantly mixed Th1 and Th17 immune responses, with high titers of specific IgG1 and IgG2a. Splenocytes of immunized mice exhibited strong antigen-specific memory responses, resulting in the secretion of high amounts of IFN-γ and IL-17, and low levels of IL-4. Immunization with rFliD caused a significant reduction in H. pylori bacterial load relative to naïve control mice (p < 0.001), demonstrating a robust protective effect. Taken together, these results suggest that subcutaneous vaccination with rFliD formulated with CpG or Addavax could be considered as a potential candidate for the development of a subunit vaccine against H. pylori infection.


Assuntos
Proteínas de Bactérias/uso terapêutico , Vacinas Bacterianas/uso terapêutico , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/isolamento & purificação , Vacinas Bacterianas/imunologia , Clonagem Molecular , Feminino , Helicobacter pylori/genética , Imunização/métodos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico
20.
Can J Physiol Pharmacol ; 96(2): 113-119, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28863272

RESUMO

Recombinant Helicobacter pylori neutrophil-activating protein fused with maltose-binding protein (rMBP-NAP), a potential TLR2 ligand, was reported to possess immunomodulatory effects on in situ tumors in our previous study. In the present work, we attempt to elucidate the effect of rMBP-NAP at the local immune modulation in B16-F10-induced metastatic lung cancer. Our results demonstrated that growth of B16-F10 melanoma metastases in the lung was significantly arrested after rMBP-NAP treatment, along with marked reduction in metastatic lung nodules and significant increase in survival. The treatment induced both local and systemic immune responses, which were associated with higher influx of CD4+/CD8+ T cells and drove toward Th1-like and cytotoxic immune environment. Moreover, rMBP-NAP also showed significant anti-angiogenic activity by reducing vascularization in lung tumor sections. rMBP-NAP could induce antitumor immunity through activating Th1 cells and producing pro-inflammatory cytokines, which are responsible for the effective cytotoxic immune response against cancer progression. Our findings indicate that rMBP-NAP might be a novel antitumor therapeutic strategy.


Assuntos
Proteínas de Bactérias/uso terapêutico , Progressão da Doença , Imunidade , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Proteínas Ligantes de Maltose/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Imunidade/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Neovascularização Patológica/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Baço/patologia , Análise de Sobrevida , Células Th1/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA